23 results on '"Michele L. Lenoue-Newton"'
Search Results
2. The My Cancer Genome clinical trial data model and trial curation workflow.
- Author
-
Neha Jain, Kathleen F. Mittendorf, Marilyn Holt, Michele L. LeNoue-Newton, Ian Maurer, Clinton Miller, Matthew Stachowiak, Michelle Botyrius, James Cole, Christine Micheel, and Mia A. Levy
- Published
- 2020
- Full Text
- View/download PDF
3. Genomic Landscapes and Hallmarks of Mutant RAS in Human Cancers
- Author
-
Robert B. Scharpf, Archana Balan, Biagio Ricciuti, Jacob Fiksel, Christopher Cherry, Chenguang Wang, Michele L. Lenoue-Newton, Hira A. Rizvi, James R. White, Alexander S. Baras, Jordan Anaya, Blair V. Landon, Marta Majcherska-Agrawal, Paola Ghanem, Jocelyn Lee, Leon Raskin, Andrew S. Park, Huakang Tu, Hil Hsu, Kathryn C. Arbour, Mark M. Awad, Gregory J. Riely, Christine M. Lovly, and Valsamo Anagnostou
- Subjects
Proto-Oncogene Proteins p21(ras) ,Cancer Research ,Lung Neoplasms ,Oncology ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Tumor Microenvironment ,Humans ,Bayes Theorem ,Genomics ,Article - Abstract
The RAS family of small GTPases represents the most commonly activated oncogenes in human cancers. To better understand the prevalence of somatic RAS mutations and the compendium of genes that are coaltered in RAS-mutant tumors, we analyzed targeted next-generation sequencing data of 607,863 mutations from 66,372 tumors in 51 cancer types in the AACR Project GENIE Registry. Bayesian hierarchical models were implemented to estimate the cancer-specific prevalence of RAS and non-RAS somatic mutations, to evaluate co-occurrence and mutual exclusivity, and to model the effects of tumor mutation burden and mutational signatures on comutation patterns. These analyses revealed differential RAS prevalence and comutations with non-RAS genes in a cancer lineage-dependent and context-dependent manner, with differences across age, sex, and ethnic groups. Allele-specific RAS co-mutational patterns included an enrichment in NTRK3 and chromatin-regulating gene mutations in KRAS G12C-mutant non–small cell lung cancer. Integrated multiomic analyses of 10,217 tumors from The Cancer Genome Atlas (TCGA) revealed distinct genotype-driven gene expression programs pointing to differential recruitment of cancer hallmarks as well as phenotypic differences and immune surveillance states in the tumor microenvironment of RAS-mutant tumors. The distinct genomic tracks discovered in RAS-mutant tumors reflected differential clinical outcomes in TCGA cohort and in an independent cohort of patients with KRAS G12C-mutant non–small cell lung cancer that received immunotherapy-containing regimens. The RAS genetic architecture points to cancer lineage–specific therapeutic vulnerabilities that can be leveraged for rationally combining RAS-mutant allele-directed therapies with targeted therapies and immunotherapy. Significance: The complex genomic landscape of RAS-mutant tumors is reflective of selection processes in a cancer lineage–specific and context-dependent manner, highlighting differential therapeutic vulnerabilities that can be clinically translated.
- Published
- 2022
- Full Text
- View/download PDF
4. Integrating Cancer Genomic Data into Decision Support at the VA: Hematologic Oncology as a Use Case.
- Author
-
Dax M. Westerman, Fern FitzHenry, Michael E. Matheny, Michele L. LeNoue-Newton, Claudio A. Mosse, Ian Maurer, Matthew Stachowiak, Justin A. Yeakley, James Cole, Regina M. Thomas, and Mia A. Levy
- Published
- 2018
5. Data from AACR Project GENIE: 100,000 Cases and Beyond
- Author
-
Brady Bernard, Shawn M. Sweeney, Thomas V. Yu, Nikolaus Schultz, Stephen-John Sammut, Andrea Ovalle, Tali Mazor, James Lindsay, Marilyn M. Li, Michele L. Lenoue-Newton, Priti Kumari, Haley Hunter-Zinck, Michelle F. Green, Matthew Galvin, Gary J. Doherty, Jeff P. Bruce, Jonathan L. Bell, and Trevor J. Pugh
- Abstract
The American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) is an international pan-cancer registry with the goal to inform cancer research and clinical care worldwide. Founded in late 2015, the milestone GENIE 9.1-public release contains data from >110,000 tumors from >100,000 people treated at 19 cancer centers from the United States, Canada, the United Kingdom, France, the Netherlands, and Spain. Here, we demonstrate the use of these real-world data, harmonized through a centralized data resource, to accurately predict enrollment on genome-guided trials, discover driver alterations in rare tumors, and identify cancer types without actionable mutations that could benefit from comprehensive genomic analysis. The extensible data infrastructure and governance framework support additional deep patient phenotyping through biopharmaceutical collaborations and expansion to include new data types such as cell-free DNA sequencing. AACR Project GENIE continues to serve a global precision medicine knowledge base of increasing impact to inform clinical decision-making and bring together cancer researchers internationally.Significance:AACR Project GENIE has now accrued data from >110,000 tumors, placing it among the largest repository of publicly available, clinically annotated genomic data in the world. GENIE has emerged as a powerful resource to evaluate genome-guided clinical trial design, uncover drivers of cancer subtypes, and inform real-world use of genomic data.This article is highlighted in the In This Issue feature, p. 2007
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table from AACR Project GENIE: 100,000 Cases and Beyond
- Author
-
Brady Bernard, Shawn M. Sweeney, Thomas V. Yu, Nikolaus Schultz, Stephen-John Sammut, Andrea Ovalle, Tali Mazor, James Lindsay, Marilyn M. Li, Michele L. Lenoue-Newton, Priti Kumari, Haley Hunter-Zinck, Michelle F. Green, Matthew Galvin, Gary J. Doherty, Jeff P. Bruce, Jonathan L. Bell, and Trevor J. Pugh
- Abstract
Supplementary Table from AACR Project GENIE: 100,000 Cases and Beyond
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure from AACR Project GENIE: 100,000 Cases and Beyond
- Author
-
Brady Bernard, Shawn M. Sweeney, Thomas V. Yu, Nikolaus Schultz, Stephen-John Sammut, Andrea Ovalle, Tali Mazor, James Lindsay, Marilyn M. Li, Michele L. Lenoue-Newton, Priti Kumari, Haley Hunter-Zinck, Michelle F. Green, Matthew Galvin, Gary J. Doherty, Jeff P. Bruce, Jonathan L. Bell, and Trevor J. Pugh
- Abstract
Supplementary Figure from AACR Project GENIE: 100,000 Cases and Beyond
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Methods, Supplemental Tables 1-2, Supplemental Figures 1-4 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
Supplemental Methods. Supplemental Table 1: ââ,¬â€¹Genomic Data Characterization by Center. Supplemental Table 2: ââ,¬â€¹Gene Panels Submitted by Each Center. Figure S1: Number of putative germline SNPs per sample, before and after uniform germline filtering. Figure S2ââ,¬â€¹. Distribution of total somatic mutation burden per sample stratified by sequencing panel. Figure S3: ââ,¬â€¹Log-scale comparison of mutation frequencies at hotspot sites between GENIE (data aggregated from all sequencing panels) and cancerhotspots.org (CHS) using a binomial test. Figure S4:ââ,¬â€¹ Comparison of mutation frequencies at hotspot sites in each GENIE sequencing panel with cancerhotspots.org (CHS) using a binomial test.
- Published
- 2023
- Full Text
- View/download PDF
9. Table S4 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
Table S4
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental File 1 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
AACR GENIE Data Guide
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data from Characteristics and Outcome of AKT1E17K-Mutant Breast Cancer Defined through AACR Project GENIE, a Clinicogenomic Registry
- Author
-
David M. Hyman, Deborah Schrag, Alexia Iasonos, Philippe L. Bedard, Funda Meric-Bernstam, Mia Levy, Fabrice André, Charles L. Sawyers, Ben H. Park, Seth Sheffler-Collins, Jocelyn Lee, Stuart M. Gardos, Andrew Zarski, Nikolaus Schultz, JianJiong Gao, Shawn M. Sweeney, Ritika Kundra, Benjamin E. Gross, Jan Hudecek, Hugo Horlings, Chetna Wathoo, Christine M. Micheel, Semih Dogan, Natalie Blauvelt, Michele L. Lenoue-Newton, Michael J. Hasset, Monica Arnedos, Eva M. Lepisto, Celeste Yu, Bastien Nguyen, Qin Zhou, and Lillian M. Smyth
- Abstract
Supplementary Tables and Figures
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Characteristics and Outcome of AKT1E17K-Mutant Breast Cancer Defined through AACR Project GENIE, a Clinicogenomic Registry
- Author
-
David M. Hyman, Deborah Schrag, Alexia Iasonos, Philippe L. Bedard, Funda Meric-Bernstam, Mia Levy, Fabrice André, Charles L. Sawyers, Ben H. Park, Seth Sheffler-Collins, Jocelyn Lee, Stuart M. Gardos, Andrew Zarski, Nikolaus Schultz, JianJiong Gao, Shawn M. Sweeney, Ritika Kundra, Benjamin E. Gross, Jan Hudecek, Hugo Horlings, Chetna Wathoo, Christine M. Micheel, Semih Dogan, Natalie Blauvelt, Michele L. Lenoue-Newton, Michael J. Hasset, Monica Arnedos, Eva M. Lepisto, Celeste Yu, Bastien Nguyen, Qin Zhou, and Lillian M. Smyth
- Abstract
AKT inhibitors have promising activity in AKT1E17K-mutant estrogen receptor (ER)–positive metastatic breast cancer, but the natural history of this rare genomic subtype remains unknown. Utilizing AACR Project GENIE, an international clinicogenomic data-sharing consortium, we conducted a comparative analysis of clinical outcomes of patients with matched AKT1E17K-mutant (n = 153) and AKT1–wild-type (n = 302) metastatic breast cancer. AKT1-mutant cases had similar adjusted overall survival (OS) compared with AKT1–wild-type controls (median OS, 24.1 vs. 29.9, respectively; P = 0.98). AKT1-mutant cases enjoyed longer durations on mTOR inhibitor therapy, an observation previously unrecognized in pivotal clinical trials due to the rarity of this alteration. Other baseline clinicopathologic features, as well as durations on other classes of therapy, were broadly similar. In summary, we demonstrate the feasibility of using a novel and publicly accessible clincogenomic registry to define outcomes in a rare genomically defined cancer subtype, an approach with broad applicability to precision oncology.Significance:We delineate the natural history of a rare genomically distinct cancer, AKT1E17K-mutant ER-positive breast cancer, using a publicly accessible registry of real-world patient data, thereby illustrating the potential to inform drug registration through synthetic control data.See related commentary by Castellanos and Baxi, p. 490.
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Author
-
Monica Arnedos, Christine M. Micheel, Fei Ye, Alshad S. Lalani, Grace Mann, Feng Xu, Lisa D. Eli, Mia Levy, Chetna Wathoo, Celeste Yu, Semih Dogan, Lillian Smyth, Fabrice Andre, Eva M. Lepisto, Deborah Schrag, Rinaa S. Punglia, Funda Meric-Bernstam, Philippe L. Bedard, Natalie Blauvelt, David M. Hyman, Thomas Stricker, Sheau-Chiann Chen, and Michele L. LeNoue-Newton
- Abstract
Purpose:We wanted to determine the prognosis and the phenotypic characteristics of hormone receptor–positive advanced breast cancer tumors harboring an ERBB2 mutation in the absence of a HER2 amplification.Experimental Design:We retrospectively collected information from the American Association of Cancer Research-Genomics Evidence Neoplasia Information Exchange registry database from patients with hormone receptor–positive, HER2-negative, ERBB2-mutated advanced breast cancer. Phenotypic and co-mutational features, as well as response to treatment and outcome were compared with matched control cases ERBB2 wild type.Results:A total of 45 ERBB2-mutant cases were identified for 90 matched controls. The presence of an ERBB2 mutation was not associated with worse outcome determined by overall survival (OS) from first metastatic relapse. No significant differences were observed in phenotypic characteristics apart from higher lobular infiltrating subtype in the ERBB2-mutated group. ERBB2 mutation did not seem to have an impact in response to treatment or time-to-progression (TTP) to endocrine therapy compared with ERBB2 wild type. In the co-mutational analyses, CDH1 mutation was more frequent in the ERBB2-mutated group (FDR < 1). Although not significant, fewer co-occurring ESR1 mutations and more KRAS mutations were identified in the ERBB2-mutated group.Conclusions:ERBB2-activating mutation was not associated with a worse OS from time of first metastatic relapse, or differences in TTP on treatment as compared with a series of matched controls. Although not significant, differences in coexisting mutations (CDH1, ESR1, and KRAS) were noted between the ERBB2-mutated and the control group.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Genomic Landscapes and Hallmarks of Mutant RAS in Human Cancers
- Author
-
Valsamo Anagnostou, Christine M. Lovly, Gregory J. Riely, Mark M. Awad, Kathryn C. Arbour, Hil Hsu, Huakang Tu, Andrew S. Park, Leon Raskin, Jocelyn Lee, Paola Ghanem, Marta Majcherska-Agrawal, Blair V. Landon, Jordan Anaya, Alexander S. Baras, James R. White, Hira A. Rizvi, Michele L. Lenoue-Newton, Chenguang Wang, Christopher Cherry, Jacob Fiksel, Biagio Ricciuti, Archana Balan, and Robert B. Scharpf
- Abstract
The RAS family of small GTPases represents the most commonly activated oncogenes in human cancers. To better understand the prevalence of somatic RAS mutations and the compendium of genes that are coaltered in RAS-mutant tumors, we analyzed targeted next-generation sequencing data of 607,863 mutations from 66,372 tumors in 51 cancer types in the AACR Project GENIE Registry. Bayesian hierarchical models were implemented to estimate the cancer-specific prevalence of RAS and non-RAS somatic mutations, to evaluate co-occurrence and mutual exclusivity, and to model the effects of tumor mutation burden and mutational signatures on comutation patterns. These analyses revealed differential RAS prevalence and comutations with non-RAS genes in a cancer lineage-dependent and context-dependent manner, with differences across age, sex, and ethnic groups. Allele-specific RAS co-mutational patterns included an enrichment in NTRK3 and chromatin-regulating gene mutations in KRAS G12C-mutant non–small cell lung cancer. Integrated multiomic analyses of 10,217 tumors from The Cancer Genome Atlas (TCGA) revealed distinct genotype-driven gene expression programs pointing to differential recruitment of cancer hallmarks as well as phenotypic differences and immune surveillance states in the tumor microenvironment of RAS-mutant tumors. The distinct genomic tracks discovered in RAS-mutant tumors reflected differential clinical outcomes in TCGA cohort and in an independent cohort of patients with KRAS G12C-mutant non–small cell lung cancer that received immunotherapy-containing regimens. The RAS genetic architecture points to cancer lineage–specific therapeutic vulnerabilities that can be leveraged for rationally combining RAS-mutant allele-directed therapies with targeted therapies and immunotherapy.Significance:The complex genomic landscape of RAS-mutant tumors is reflective of selection processes in a cancer lineage–specific and context-dependent manner, highlighting differential therapeutic vulnerabilities that can be clinically translated.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Data from Genomic Landscapes and Hallmarks of Mutant RAS in Human Cancers
- Author
-
Valsamo Anagnostou, Christine M. Lovly, Gregory J. Riely, Mark M. Awad, Kathryn C. Arbour, Hil Hsu, Huakang Tu, Andrew S. Park, Leon Raskin, Jocelyn Lee, Paola Ghanem, Marta Majcherska-Agrawal, Blair V. Landon, Jordan Anaya, Alexander S. Baras, James R. White, Hira A. Rizvi, Michele L. Lenoue-Newton, Chenguang Wang, Christopher Cherry, Jacob Fiksel, Biagio Ricciuti, Archana Balan, and Robert B. Scharpf
- Abstract
Supplementary Data from Genomic Landscapes and Hallmarks of Mutant RAS in Human Cancers
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Author
-
Monica Arnedos, Christine M. Micheel, Fei Ye, Alshad S. Lalani, Grace Mann, Feng Xu, Lisa D. Eli, Mia Levy, Chetna Wathoo, Celeste Yu, Semih Dogan, Lillian Smyth, Fabrice Andre, Eva M. Lepisto, Deborah Schrag, Rinaa S. Punglia, Funda Meric-Bernstam, Philippe L. Bedard, Natalie Blauvelt, David M. Hyman, Thomas Stricker, Sheau-Chiann Chen, and Michele L. LeNoue-Newton
- Abstract
Supplementary Figure from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Author
-
Monica Arnedos, Christine M. Micheel, Fei Ye, Alshad S. Lalani, Grace Mann, Feng Xu, Lisa D. Eli, Mia Levy, Chetna Wathoo, Celeste Yu, Semih Dogan, Lillian Smyth, Fabrice Andre, Eva M. Lepisto, Deborah Schrag, Rinaa S. Punglia, Funda Meric-Bernstam, Philippe L. Bedard, Natalie Blauvelt, David M. Hyman, Thomas Stricker, Sheau-Chiann Chen, and Michele L. LeNoue-Newton
- Abstract
Supplementary Table from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Published
- 2023
- Full Text
- View/download PDF
18. Allele-specific activation, enzyme kinetics, and inhibitor sensitivities of EGFR exon 19 deletion mutations in lung cancer
- Author
-
Benjamin P. Brown, Yun-Kai Zhang, Soyeon Kim, Patrick Finneran, Yingjun Yan, Zhenfang Du, Jiyoon Kim, Abigail Leigh Hartzler, Michele L. LeNoue-Newton, Adam W. Smith, Jens Meiler, and Christine M. Lovly
- Subjects
Enzyme Activation ,ErbB Receptors ,Kinetics ,Lung Neoplasms ,Multidisciplinary ,Carcinoma, Non-Small-Cell Lung ,Amino Acid Motifs ,Humans ,Exons ,Neoplasm Recurrence, Local ,Protein Kinase Inhibitors ,Alleles ,Sequence Deletion - Abstract
Oncogenic mutations within the epidermal growth factor receptor (EGFR) are found in 15-30% of all non-small cell lung carcinomas. The term exon 19 deletion (ex19del) is collectively used to refer to more than 20 distinct genomic alterations within exon 19 that comprise the most common EGFR mutation subtype in lung cancer. Despite this heterogeneity, clinical treatment decisions are made irrespective of which EGFR ex19del variant is present within the tumor, and there is a paucity of information regarding how individual ex19del variants influence protein structure and function. Herein, we identify allele-specific functional differences among ex19del variants attributable to recurring sequence and structure motifs. We build all-atom structural models of 60 ex19del variants identified in patients and combine ~400 μs of molecular dynamics simulations with biochemical and biophysical experiments to analyze three ex19del mutations (E746_A750, E746_S752>V, and L747_A750>P). We demonstrate that sequence variation in ex19del alters oncogenic cell growth, dimerization propensity, and tyrosine kinase inhibitor (TKI) sensitivity. We show that in contrast to E746_A750 and E746_S752>V, the L747_A750>P variant forms highly active ligand-independent dimers. E746_S752>V displays the least TKI sensitivity among the variants tested, which enzyme kinetic analysis and TKI inhibition experiments suggest is due to increased ATP Km relative to the common E746_A750 variant. Through these analyses, we propose an expanded framework for interpreting ex19del variants and new considerations for therapeutic intervention.SignificanceEGFR mutations are detected in approximately 30% of all lung adenocarcinomas, and the most common EGFR mutation occurring in ~50% of patients is termed “exon 19 deletion” (ex19del). Despite the existence of dozens of different genomic variants comprising what is generically referred to clinically as ex19del, clinicians currently do not distinguish between ex19del variants in considering treatment options, and the differences between ex19del variants are largely unstudied in the broader scientific community. Herein, we describe functional differences between distinct EGFR ex19del variants attributable to the structural features of each variant. These findings suggest a possible explanation for observed differences in patient outcomes stratified by ex19del subtype and reinforce the need for allele-specific considerations in clinical treatment decision making.
- Published
- 2022
- Full Text
- View/download PDF
19. AACR Project GENIE: 100,000 Cases and Beyond
- Author
-
Trevor J, Pugh, Jonathan L, Bell, Jeff P, Bruce, Gary J, Doherty, Matthew, Galvin, Michelle F, Green, Haley, Hunter-Zinck, Priti, Kumari, Michele L, Lenoue-Newton, Marilyn M, Li, James, Lindsay, Tali, Mazor, Andrea, Ovalle, Stephen-John, Sammut, Nikolaus, Schultz, Thomas V, Yu, Shawn M, Sweeney, and Brady, Bernard
- Subjects
Oncology ,Neoplasms ,Mutation ,Humans ,Genomics ,Precision Medicine ,Cell-Free Nucleic Acids ,United States - Abstract
The American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) is an international pan-cancer registry with the goal to inform cancer research and clinical care worldwide. Founded in late 2015, the milestone GENIE 9.1-public release contains data from >110,000 tumors from >100,000 people treated at 19 cancer centers from the United States, Canada, the United Kingdom, France, the Netherlands, and Spain. Here, we demonstrate the use of these real-world data, harmonized through a centralized data resource, to accurately predict enrollment on genome-guided trials, discover driver alterations in rare tumors, and identify cancer types without actionable mutations that could benefit from comprehensive genomic analysis. The extensible data infrastructure and governance framework support additional deep patient phenotyping through biopharmaceutical collaborations and expansion to include new data types such as cell-free DNA sequencing. AACR Project GENIE continues to serve a global precision medicine knowledge base of increasing impact to inform clinical decision-making and bring together cancer researchers internationally. Significance: AACR Project GENIE has now accrued data from >110,000 tumors, placing it among the largest repository of publicly available, clinically annotated genomic data in the world. GENIE has emerged as a powerful resource to evaluate genome-guided clinical trial design, uncover drivers of cancer subtypes, and inform real-world use of genomic data. This article is highlighted in the In This Issue feature, p. 2007
- Published
- 2021
20. Natural History and Characteristics of ERBB2-mutated Hormone Receptor-positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case-control Study from AACR Project GENIE
- Author
-
Michele L. LeNoue-Newton, Sheau-Chiann Chen, Thomas Stricker, David M. Hyman, Natalie Blauvelt, Philippe L. Bedard, Funda Meric-Bernstam, Rinaa S. Punglia, Deborah Schrag, Eva M. Lepisto, Fabrice Andre, Lillian Smyth, Semih Dogan, Celeste Yu, Chetna Wathoo, Mia Levy, Lisa D. Eli, Feng Xu, Grace Mann, Alshad S. Lalani, Fei Ye, Christine M. Micheel, and Monica Arnedos
- Subjects
Proto-Oncogene Proteins p21(ras) ,Cancer Research ,Carcinoma, Lobular ,Oncology ,Receptor, ErbB-2 ,Recurrence ,Case-Control Studies ,Mutation ,Biomarkers, Tumor ,Humans ,Breast Neoplasms ,Female ,Retrospective Studies - Abstract
Purpose: We wanted to determine the prognosis and the phenotypic characteristics of hormone receptor–positive advanced breast cancer tumors harboring an ERBB2 mutation in the absence of a HER2 amplification. Experimental Design: We retrospectively collected information from the American Association of Cancer Research-Genomics Evidence Neoplasia Information Exchange registry database from patients with hormone receptor–positive, HER2-negative, ERBB2-mutated advanced breast cancer. Phenotypic and co-mutational features, as well as response to treatment and outcome were compared with matched control cases ERBB2 wild type. Results: A total of 45 ERBB2-mutant cases were identified for 90 matched controls. The presence of an ERBB2 mutation was not associated with worse outcome determined by overall survival (OS) from first metastatic relapse. No significant differences were observed in phenotypic characteristics apart from higher lobular infiltrating subtype in the ERBB2-mutated group. ERBB2 mutation did not seem to have an impact in response to treatment or time-to-progression (TTP) to endocrine therapy compared with ERBB2 wild type. In the co-mutational analyses, CDH1 mutation was more frequent in the ERBB2-mutated group (FDR < 1). Although not significant, fewer co-occurring ESR1 mutations and more KRAS mutations were identified in the ERBB2-mutated group. Conclusions: ERBB2-activating mutation was not associated with a worse OS from time of first metastatic relapse, or differences in TTP on treatment as compared with a series of matched controls. Although not significant, differences in coexisting mutations (CDH1, ESR1, and KRAS) were noted between the ERBB2-mutated and the control group.
- Published
- 2021
21. American Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange: From Inception to First Data Release and Beyond—Lessons Learned and Member Institutions’ Perspectives
- Author
-
Shawn M. Sweeney, Gerrit A. Meijer, Michele L. LeNoue-Newton, Christine M. Micheel, Philippe L. Bedard, Victor E. Velculescu, Charles L. Sawyers, Nikolaus Schultz, Mia A. Levy, Justin Guinney, Fabrice Andre, Barrett J. Rollins, and Kenna R. Mills Shaw
- Subjects
0301 basic medicine ,Association (object-oriented programming) ,media_common.quotation_subject ,Genomics ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Political science ,Institution ,Humans ,Precision Medicine ,Project management ,Intersectoral Collaboration ,Review Articles ,Societies, Medical ,Information exchange ,media_common ,Information Dissemination ,business.industry ,Data Collection ,General Medicine ,16. Peace & justice ,United States ,3. Good health ,030104 developmental biology ,Precision oncology ,030220 oncology & carcinogenesis ,Informatics ,Cancer research ,business ,Data release - Abstract
The American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) is an international data-sharing consortium focused on enabling advances in precision oncology through the gathering and sharing of tumor genetic sequencing data linked with clinical data. The project’s history, operational structure, lessons learned, and institutional perspectives on participation in the data-sharing consortium are reviewed. Individuals involved with the inception and execution of AACR Project GENIE from each member institution described their experiences and lessons learned. The consortium was conceived in January 2014 and publicly released its first data set in January 2017, which consisted of 18,804 samples from 18,324 patients contributed by the eight founding institutions. Commitment and contributions from many individuals at AACR and the member institutions were crucial to the consortium’s success. These individuals filled leadership, project management, informatics, data curation, contracts, ethics, and security roles. Many lessons were learned during the first 3 years of the consortium, including on how to gather, harmonize, and share data; how to make decisions and foster collaboration; and how to set the stage for continued participation and expansion of the consortium. We hope that the lessons shared here will assist new GENIE members as well as others who embark on the journey of forming a genomic data–sharing consortium.
- Published
- 2018
- Full Text
- View/download PDF
22. Characteristics and Outcome of
- Author
-
Lillian M, Smyth, Qin, Zhou, Bastien, Nguyen, Celeste, Yu, Eva M, Lepisto, Monica, Arnedos, Michael J, Hasset, Michele L, Lenoue-Newton, Natalie, Blauvelt, Semih, Dogan, Christine M, Micheel, Chetna, Wathoo, Hugo, Horlings, Jan, Hudecek, Benjamin E, Gross, Ritika, Kundra, Shawn M, Sweeney, JianJiong, Gao, Nikolaus, Schultz, Andrew, Zarski, Stuart M, Gardos, Jocelyn, Lee, Seth, Sheffler-Collins, Ben H, Park, Charles L, Sawyers, Fabrice, André, Mia, Levy, Funda, Meric-Bernstam, Philippe L, Bedard, Alexia, Iasonos, Deborah, Schrag, and David M, Hyman
- Subjects
Adult ,Aged, 80 and over ,Treatment Outcome ,Mutation ,Humans ,Breast Neoplasms ,Female ,Registries ,Middle Aged ,Proto-Oncogene Proteins c-akt ,Aged - Abstract
AKT inhibitors have promising activity in
- Published
- 2019
23. American Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange: From Inception to First Data Release and Beyond-Lessons Learned and Member Institutions' Perspectives.
- Author
-
Micheel CM, Sweeney SM, LeNoue-Newton ML, André F, Bedard PL, Guinney J, Meijer GA, Rollins BJ, Sawyers CL, Schultz N, Shaw KRM, Velculescu VE, and Levy MA
- Subjects
- Data Collection, Humans, Information Dissemination, Intersectoral Collaboration, Precision Medicine, Societies, Medical, United States, Genomics methods, Neoplasms genetics
- Abstract
The American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) is an international data-sharing consortium focused on enabling advances in precision oncology through the gathering and sharing of tumor genetic sequencing data linked with clinical data. The project's history, operational structure, lessons learned, and institutional perspectives on participation in the data-sharing consortium are reviewed. Individuals involved with the inception and execution of AACR Project GENIE from each member institution described their experiences and lessons learned. The consortium was conceived in January 2014 and publicly released its first data set in January 2017, which consisted of 18,804 samples from 18,324 patients contributed by the eight founding institutions. Commitment and contributions from many individuals at AACR and the member institutions were crucial to the consortium's success. These individuals filled leadership, project management, informatics, data curation, contracts, ethics, and security roles. Many lessons were learned during the first 3 years of the consortium, including on how to gather, harmonize, and share data; how to make decisions and foster collaboration; and how to set the stage for continued participation and expansion of the consortium. We hope that the lessons shared here will assist new GENIE members as well as others who embark on the journey of forming a genomic data-sharing consortium.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.